Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Hepatol Commun ; 8(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38619429

ABSTRACT

BACKGROUND: Mutations in the gene MTARC1 (mitochondrial amidoxime-reducing component 1) protect carriers from metabolic dysfunction-associated steatohepatitis (MASH) and cirrhosis. MTARC1 encodes the mARC1 enzyme, which is localized to the mitochondria and has no known MASH-relevant molecular function. Our studies aimed to expand on the published human genetic mARC1 data and to observe the molecular effects of mARC1 modulation in preclinical MASH models. METHODS AND RESULTS: We identified a novel human structural variant deletion in MTARC1, which is associated with various biomarkers of liver health, including alanine aminotransferase levels. Phenome-wide Mendelian Randomization analyses additionally identified novel putatively causal associations between MTARC1 expression, and esophageal varices and cardiorespiratory traits. We observed that protective MTARC1 variants decreased protein accumulation in in vitro overexpression systems and used genetic tools to study mARC1 depletion in relevant human and mouse systems. Hepatocyte mARC1 knockdown in murine MASH models reduced body weight, liver steatosis, oxidative stress, cell death, and fibrogenesis markers. mARC1 siRNA treatment and overexpression modulated lipid accumulation and cell death consistently in primary human hepatocytes, hepatocyte cell lines, and primary human adipocytes. mARC1 depletion affected the accumulation of distinct lipid species and the expression of inflammatory and mitochondrial pathway genes/proteins in both in vitro and in vivo models. CONCLUSIONS: Depleting hepatocyte mARC1 improved metabolic dysfunction-associated steatotic liver disease-related outcomes. Given the functional role of mARC1 in human adipocyte lipid accumulation, systemic targeting of mARC1 should be considered when designing mARC1 therapies. Our data point to plasma lipid biomarkers predictive of mARC1 abundance, such as Ceramide 22:1. We propose future areas of study to describe the precise molecular function of mARC1, including lipid trafficking and subcellular location within or around the mitochondria and endoplasmic reticulum.


Subject(s)
Fatty Liver , Hepatocytes , Animals , Humans , Mice , Adipocytes , Biomarkers , Ceramides , Mendelian Randomization Analysis
2.
Science ; 372(6543): 716-721, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33986176

ABSTRACT

Transcription and metabolism both influence cell function, but dedicated transcriptional control of metabolic pathways that regulate cell fate has rarely been defined. We discovered, using a chemical suppressor screen, that inhibition of the pyrimidine biosynthesis enzyme dihydroorotate dehydrogenase (DHODH) rescues erythroid differentiation in bloodless zebrafish moonshine (mon) mutant embryos defective for transcriptional intermediary factor 1 gamma (tif1γ). This rescue depends on the functional link of DHODH to mitochondrial respiration. The transcription elongation factor TIF1γ directly controls coenzyme Q (CoQ) synthesis gene expression. Upon tif1γ loss, CoQ levels are reduced, and a high succinate/α-ketoglutarate ratio leads to increased histone methylation. A CoQ analog rescues mon's bloodless phenotype. These results demonstrate that mitochondrial metabolism is a key output of a lineage transcription factor that drives cell fate decisions in the early blood lineage.


Subject(s)
Erythropoiesis , Mitochondria/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Zebrafish Proteins/metabolism , Animals , Citric Acid Cycle , DNA Methylation , Dihydroorotate Dehydrogenase , Electron Transport , Embryo, Nonmammalian/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Histones/metabolism , Leflunomide/pharmacology , Metabolic Networks and Pathways , Methylation , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxygen Consumption , Transcription Factors/genetics , Ubiquinone/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Curr Protoc Pharmacol ; 91(1): e81, 2020 12.
Article in English | MEDLINE | ID: mdl-33332748

ABSTRACT

To assess the role of a protein, protein loss phenotypic studies can be used, most commonly through mutagenesis RNAi or CRISPR knockout. Such studies have been critical for the understanding of protein function and the identification of putative therapeutic targets for numerous human disease states. However, these methodological approaches present challenges because they are not easily reversible, and if an essential gene is targeted, an associated loss of cell viability can potentially hinder further studies. Here we present a reversible and conditional live-cell knockout strategy that is applicable to numerous proteins. This modular protein-tagging approach regulates target loss at the protein, rather than the genomic, level through the use of HaloPROTAC3, which specifically degrades HaloTag fusion proteins via recruitment of the VHL E3 ligase component. To enable HaloTag-mediated degradation of endogenous proteins, we provide protocols for HaloTag genomic insertion at the protein N or C terminus via CRISPR/Cas9 and use of HaloTag fluorescent ligands to enrich edited cells via Fluorescence-Activated Cell Sorting (FACS). Using these approaches, endogenous HaloTag fusion proteins present in various subcellular locations can be degraded by HaloPROTAC3. As detecting the degradation of endogenous targets is challenging, the 11-amino-acid peptide tag HiBiT is added to the HaloTag fusion to allows the sensitive luminescence detection of HaloTag fusion levels without the use of antibodies. Lastly, we demonstrate, through comparison of HaloPROTAC3 degradation with that of another fusion tag PROTAC, dTAG-13, that HaloPROTAC3 has a faster degradation rate and similar extent of degradation. © 2020 The Authors. Basic Protocol 1: CRISPR/Cas9 insertion of HaloTag or HiBiT-HaloTag Basic Protocol 2: HaloPROTAC3 degradation of endogenous HaloTag fusions.


Subject(s)
CRISPR-Cas Systems , Proteolysis , Recombinant Fusion Proteins/chemistry , Cell Line , Electroporation , Humans
4.
Lab Chip ; 20(22): 4152-4165, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33034335

ABSTRACT

Adipose is a distributed organ that performs vital endocrine and energy homeostatic functions. Hypertrophy of white adipocytes is a primary mode of both adaptive and maladaptive weight gain in animals and predicts metabolic syndrome independent of obesity. Due to the failure of conventional culture to recapitulate adipocyte hypertrophy, technology for production of adult-size adipocytes would enable applications such as in vitro testing of weight loss therapeutics. To model adaptive adipocyte hypertrophy in vitro, we designed and built fat-on-a-chip using fiber networks inspired by extracellular matrix in adipose tissue. Fiber networks extended the lifespan of differentiated adipocytes, enabling growth to adult sizes. By micropatterning preadipocytes in a native cytoarchitecture and by adjusting cell-to-cell spacing, rates of hypertrophy were controlled independent of culture time or differentiation efficiency. In vitro hypertrophy followed a nonlinear, nonexponential growth model similar to human development and elicited transcriptomic changes that increased overall similarity with primary tissue. Cells on the chip responded to simulated meals and starvation, which potentiated some adipocyte endocrine and metabolic functions. To test the utility of the platform for therapeutic development, transcriptional network analysis was performed, and retinoic acid receptors were identified as candidate drug targets. Regulation by retinoid signaling was suggested further by pharmacological modulation, where activation accelerated and inhibition slowed hypertrophy. Altogether, this work presents technology for mature adipocyte engineering, addresses the regulation of cell growth, and informs broader applications for synthetic adipose in pharmaceutical development, regenerative medicine, and cellular agriculture.


Subject(s)
Adipocytes, White , Fasting , Adipose Tissue , Adult , Animals , Humans , Hypertrophy , Obesity
5.
Stem Cell Reports ; 14(4): 590-602, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32243843

ABSTRACT

We previously discovered in mouse adipocytes an lncRNA (the homolog of human LINC00116) regulating adipogenesis that contains a highly conserved coding region. Here, we show human protein expression of a peptide within LINC00116, and demonstrate that this peptide modulates triglyceride clearance in human adipocytes by regulating lipolysis and mitochondrial ß-oxidation. This gene has previously been identified as mitoregulin (MTLN). We conclude that MTLN has a regulatory role in adipocyte metabolism as demonstrated by systemic lipid phenotypes in knockout mice. We also assert its adipocyte-autonomous phenotypes in both isolated murine adipocytes as well as human stem cell-derived adipocytes. MTLN directly interacts with the ß subunit of the mitochondrial trifunctional protein, an enzyme critical in the ß-oxidation of long-chain fatty acids. Our human and murine models contend that MTLN could be an avenue for further therapeutic research, albeit not without caveats, for example, by promoting white adipocyte triglyceride clearance in obese subjects.


Subject(s)
Adipocytes/metabolism , Mitochondrial Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Respiration , Conserved Sequence , Energy Metabolism , Humans , Lipid Metabolism , Lipids/blood , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/chemistry , Oxidation-Reduction
6.
Circ Res ; 126(3): 330-346, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31739742

ABSTRACT

Rationale: Genome-wide association studies have identified genetic loci associated with insulin resistance (IR) but pinpointing the causal genes of a risk locus has been challenging. Objective: To identify candidate causal genes for IR, we screened regional and biologically plausible genes (16 in total) near the top 10 IR-loci in risk-relevant cell types, namely preadipocytes and adipocytes. Methods and Results: We generated 16 human Simpson-Golabi-Behmel syndrome preadipocyte knockout lines each with a single IR-gene knocked out by lentivirus-mediated CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 system. We evaluated each gene knockout by screening IR-relevant phenotypes in the 3 insulin-sensitizing mechanisms, including adipogenesis, lipid metabolism, and insulin signaling. We performed genetic analyses using data on the genotype-tissue expression portal expression quantitative trait loci database and accelerating medicines partnership type 2 diabetes mellitus Knowledge Portal to evaluate whether candidate genes prioritized by our in vitro studies were expression quantitative trait loci genes in human subcutaneous adipose tissue, and whether expression of these genes is associated with risk of IR, type 2 diabetes mellitus, and cardiovascular diseases. We further validated the functions of 3 new adipose IR genes by overexpression-based phenotypic rescue in the Simpson-Golabi-Behmel syndrome preadipocyte knockout lines. Twelve genes, PPARG, IRS-1, FST, PEPD, PDGFC, MAP3K1, GRB14, ARL15, ANKRD55, RSPO3, COBLL1, and LYPLAL1, showed diverse phenotypes in the 3 insulin-sensitizing mechanisms, and the first 7 of these genes could affect all the 3 mechanisms. Five out of 6 expression quantitative trait loci genes are among the top candidate causal genes and the abnormal expression levels of these genes (IRS-1, GRB14, FST, PEPD, and PDGFC) in human subcutaneous adipose tissue could be associated with increased risk of IR, type 2 diabetes mellitus, and cardiovascular disease. Phenotypic rescue by overexpression of the candidate causal genes (FST, PEPD, and PDGFC) in the Simpson-Golabi-Behmel syndrome preadipocyte knockout lines confirmed their function in adipose IR. Conclusions: Twelve genes showed diverse phenotypes indicating differential roles in insulin sensitization, suggesting mechanisms bridging the association of their genomic loci with IR. We prioritized PPARG, IRS-1, GRB14, MAP3K1, FST, PEPD, and PDGFC as top candidate genes. Our work points to novel roles for FST, PEPD, and PDGFC in adipose tissue, with consequences for cardiometabolic diseases.


Subject(s)
Adipocytes/metabolism , Insulin Resistance/genetics , Quantitative Trait Loci , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Dipeptidases/genetics , Follistatin/genetics , Genome-Wide Association Study/methods , Humans , Insulin Receptor Substrate Proteins/genetics , Loss of Function Mutation , Lymphokines/genetics , MAP Kinase Kinase Kinase 1/genetics , PPAR gamma/genetics , Platelet-Derived Growth Factor/genetics
7.
Trends Cell Biol ; 28(1): 46-57, 2018 01.
Article in English | MEDLINE | ID: mdl-29054332

ABSTRACT

Induced pluripotent stem cells (iPSCs) are powerful tools for investigating the relationship between genotype and phenotype. Recent publications have described iPSC cohort studies of common genetic variants and their effects on gene expression and cellular phenotypes. These in vitro quantitative trait locus (QTL) studies are the first experiments in a new paradigm with great potential: iPSC-based functional population genetic studies. iPSC collections from large cohorts are currently under development to facilitate the next wave of these studies, which have the potential to discover the effects of common genetic variants on cellular phenotypes and to uncover the molecular basis of common genetic diseases. Here, we describe the recent advances in this developing field, and provide a road map for future in vitro functional population genetic studies and trial-in-a-dish experiments.


Subject(s)
Genetics, Population , Genome-Wide Association Study , Induced Pluripotent Stem Cells/cytology , Pharmacogenetics , Quantitative Trait Loci , Animals , Cellular Reprogramming , Genetic Variation , Genotype , Humans , Phenotype
8.
Cell Stem Cell ; 20(4): 431-433, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388427

ABSTRACT

The National Heart, Lung, and Blood Institute's Next Generation Genetic Association Studies Consortium has used induced pluripotent stem cell technology to study the effects of common genetic variants in vitro. This issue of Cell Stem Cell and affiliated journals include several manuscripts describing the results of the consortium's efforts.


Subject(s)
Genetics, Population , Genome-Wide Association Study , Cardiovascular Diseases/pathology , Cell Differentiation , Cellular Reprogramming , Gene Expression Regulation , Genetic Variation , Humans , Induced Pluripotent Stem Cells/cytology , Phenotype
9.
Cell Stem Cell ; 20(4): 547-557.e7, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388431

ABSTRACT

Genome-wide association studies (GWAS) have highlighted a large number of genetic variants with potential disease association, but functional analysis remains a challenge. Here we describe an approach to functionally validate identified variants through differentiation of induced pluripotent stem cells (iPSCs) to study cellular pathophysiology. We collected peripheral blood cells from Framingham Heart Study participants and reprogrammed them to iPSCs. We then differentiated 68 iPSC lines into hepatocytes and adipocytes to investigate the effect of the 1p13 rs12740374 variant on cardiometabolic disease phenotypes via transcriptomics and metabolomic signatures. We observed a clear association between rs12740374 and lipid accumulation and gene expression in differentiated hepatocytes, in particular, expression of SORT1, CELSR2, and PSRC1, consistent with previous analyses of this variant using other approaches. Initial investigation of additional SNPs also highlighted correlations with gene expression. These findings suggest that iPSC-based population studies hold promise as tools for the functional validation of GWAS variants.


Subject(s)
Cell Differentiation/genetics , Genome-Wide Association Study , Induced Pluripotent Stem Cells/cytology , Metabolic Diseases/genetics , Adipocytes, White/cytology , Adipocytes, White/metabolism , Cellular Reprogramming/genetics , Chromosomes, Human, Pair 1/genetics , Cohort Studies , Down-Regulation/genetics , Genotype , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Lipid Metabolism/genetics , Metabolomics , Models, Genetic , Phenotype , Quantitative Trait Loci/genetics , Reproducibility of Results , Sequence Analysis, RNA , Tissue Donors , Transcriptome/genetics
10.
Trends Cell Biol ; 26(10): 745-755, 2016 10.
Article in English | MEDLINE | ID: mdl-27268909

ABSTRACT

The properties of tissue-specific microenvironments vary widely in the human body and demonstrably influence the structure and function of many cell types. Adipocytes are no exception, responding to cues in specialized niches to perform vital metabolic and endocrine functions. The adipose microenvironment is remodeled during tissue expansion to maintain the structural and functional integrity of the tissue and disrupted remodeling in obesity contributes to the progression of metabolic syndrome, breast cancer, and other malignancies. The increasing incidence of these obesity-related diseases and the recent focus on improved in vitro models of human tissue biology underscore growing interest in the regulatory role of adipocyte microenvironments in health and disease.


Subject(s)
Adipocytes/cytology , Cell Lineage , Cellular Microenvironment , Animals , Cell-Matrix Junctions/metabolism , Disease , Humans , Mechanotransduction, Cellular
11.
Biochem Biophys Res Commun ; 476(4): 487-492, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27246738

ABSTRACT

Type 2 diabetes follows a well-defined progressive pathogenesis, beginning with insulin resistance in metabolic tissues such as the adipose. Intracellular signaling downstream of insulin receptor activation regulates critical metabolic functions of adipose tissue, including glucose uptake, lipogenesis, lipolysis and adipokine secretion. Previous studies have used the aP2 promoter to drive Cre recombinase expression in adipose tissue. Insulin receptor (IR) knockout mice created using this aP2-Cre strategy (FIRKO mice) were protected from obesity and glucose intolerance. Later studies demonstrated the promiscuity of the aP2 promoter, casting doubts upon the tissue specificity of aP2-Cre models. It is our goal to use the increased precision of the Adipoq promoter to investigate adipocyte-specific IR function. Towards this end we generated an adipocyte-specific IR knockout (AIRKO) mouse using an Adipoq-driven Cre recombinase. Here we report AIRKO mice are less insulin sensitive throughout life, and less glucose tolerant than wild-type (WT) littermates at the age of 16 weeks. In contrast to WT littermates, the insulin sensitivity of AIRKO mice is unaffected by age or dietary regimen. At any age, AIRKO mice are comparably insulin resistant to old or obese WT mice and have a significantly reduced lifespan. Similar results were obtained when these phenotypes were re-examined in FIRKO mice. We also found that the AIRKO mouse is protected from high-fat diet-induced weight gain, corresponding with a 90% reduction in tissue weight of major adipose depots compared to WT littermates. Adipose tissue mass reduction is accompanied by hepatomegaly and increased hepatic steatosis. These data indicate that adipocyte IR function is crucial to systemic energy metabolism and has profound effects on adiposity, hepatic homeostasis and lifespan.


Subject(s)
Adipocytes/metabolism , Receptor, Insulin/metabolism , Adipose Tissue/anatomy & histology , Adipose Tissue/metabolism , Aging/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Female , Insulin Resistance/physiology , Longevity/physiology , Male , Mice , Mice, Knockout , Receptor, Insulin/deficiency , Receptor, Insulin/genetics , Signal Transduction
12.
Development ; 143(9): 1475-81, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27143754

ABSTRACT

Hepatocyte-like cells (HLCs) are derived from human pluripotent stem cells (hPSCs) in vitro, but differentiation protocols commonly give rise to a heterogeneous mixture of cells. This variability confounds the evaluation of in vitro functional assays performed using HLCs. Increased differentiation efficiency and more accurate approximation of the in vivo hepatocyte gene expression profile would improve the utility of hPSCs. Towards this goal, we demonstrate the purification of a subpopulation of functional HLCs using the hepatocyte surface marker asialoglycoprotein receptor 1 (ASGR1). We analyzed the expression profile of ASGR1-positive cells by microarray, and tested their ability to perform mature hepatocyte functions (albumin and urea secretion, cytochrome activity). By these measures, ASGR1-positive HLCs are enriched for the gene expression profile and functional characteristics of primary hepatocytes compared with unsorted HLCs. We have demonstrated that ASGR1-positive sorting isolates a functional subpopulation of HLCs from among the heterogeneous cellular population produced by directed differentiation.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Embryonic Stem Cells/cytology , Flow Cytometry/methods , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/cytology , Albumins/metabolism , Cell Differentiation/physiology , Cell Line , Cell Membrane/metabolism , Cytochromes/metabolism , Humans , Urea/metabolism
13.
Oncotarget ; 7(12): 14871-84, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26918940

ABSTRACT

Transmembrane mucins (TMs) are restricted to the apical surface of normal epithelia. In cancer, TMs not only are over-expressed, but also lose polarized distribution. MUC16/CA125 is a high molecular weight TM carrying the CA125 epitope, a well-known molecular marker for human cancers. MUC16 mRNA and protein expression was mildly stimulated by low concentrations of TNFα (2.5 ng/ml) or IFNγ (20 IU/ml) when used alone; however, combined treatment with both cytokines resulted in a moderate (3-fold or less) to large (> 10-fold) stimulation of MUC16 mRNA and protein expression in a variety of cancer cell types indicating that this may be a general response. Human cancer tissue microarray analysis indicated that MUC16 expression directly correlates with TNFα and IFNγ staining intensities in certain cancers. We show that NFκB is an important mediator of cytokine stimulation of MUC16 since siRNA-mediated knockdown of NFκB/p65 greatly reduced cytokine responsiveness. Finally, we demonstrate that the 250 bp proximal promoter region of MUC16 contains an NFκB binding site that accounts for a large portion of the TNFα response. Developing methods to manipulate MUC16 expression could provide new approaches to treating cancers whose growth or metastasis is characterized by elevated levels of TMs, including MUC16.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , CA-125 Antigen/metabolism , Endometrial Neoplasms/metabolism , Interferon-gamma/pharmacology , Ovarian Neoplasms/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Antiviral Agents/pharmacology , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , CA-125 Antigen/genetics , Endometrial Neoplasms/drug therapy , Female , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Ovarian Neoplasms/drug therapy , Protein Binding , Tumor Cells, Cultured
14.
Cell Stem Cell ; 18(1): 53-65, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26748756

ABSTRACT

Human pluripotent stem cells (hPSCs) with knockout or mutant alleles can be generated using custom-engineered nucleases. Transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 nucleases are the most commonly employed technologies for editing hPSC genomes. In this Protocol Review, we provide a brief overview of custom-engineered nucleases in the context of gene editing in hPSCs with a focus on the application of TALENs and CRISPR/Cas9. We will highlight the advantages and disadvantages of each method and discuss theoretical and technical considerations for experimental design.


Subject(s)
CRISPR-Cas Systems , Genetic Techniques , Genome, Human , Pluripotent Stem Cells/cytology , Alleles , DNA/analysis , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Genetic Vectors , Genotype , Humans , Mutation , Point Mutation , Sequence Homology, Nucleic Acid
15.
Cell Metab ; 22(2): 202-3, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26244930

ABSTRACT

Type 1 diabetes is an autoimmune disease that manifests as impaired insulin secretion, with compounding complications over time. Bhatt et al. (2015) investigate protective mechanisms in survivors of type 1 diabetes by using induced pluripotent stem cells as genetic models, uncovering novel interactions between microRNA and the DNA damage checkpoint pathway.


Subject(s)
Cell Cycle Checkpoints , DNA Damage , Diabetes Mellitus, Type 1/metabolism , Gene Expression Regulation , MicroRNAs/biosynthesis , Models, Biological , Female , Humans , Male
16.
Nat Cell Biol ; 17(8): 994-1003, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26214132

ABSTRACT

The use of human pluripotent stem cells for in vitro disease modelling and clinical applications requires protocols that convert these cells into relevant adult cell types. Here, we report the rapid and efficient differentiation of human pluripotent stem cells into vascular endothelial and smooth muscle cells. We found that GSK3 inhibition and BMP4 treatment rapidly committed pluripotent cells to a mesodermal fate and subsequent exposure to VEGF-A or PDGF-BB resulted in the differentiation of either endothelial or vascular smooth muscle cells, respectively. Both protocols produced mature cells with efficiencies exceeding 80% within six days. On purification to 99% via surface markers, endothelial cells maintained their identity, as assessed by marker gene expression, and showed relevant in vitro and in vivo functionality. Global transcriptional and metabolomic analyses confirmed that the cells closely resembled their in vivo counterparts. Our results suggest that these cells could be used to faithfully model human disease.


Subject(s)
Cell Differentiation , Cell Lineage , Endothelial Cells/physiology , Induced Pluripotent Stem Cells/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Becaplermin , Biomarkers/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/transplantation , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Human Umbilical Vein Endothelial Cells/physiology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/transplantation , Metabolomics/methods , Mice, Inbred NOD , Mice, SCID , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/transplantation , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , Phenotype , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-sis/pharmacology , Time Factors , Transcription, Genetic , Transfection , Vascular Endothelial Growth Factor A/pharmacology , Wnt Signaling Pathway/drug effects
17.
PLoS One ; 10(4): e0124578, 2015.
Article in English | MEDLINE | ID: mdl-25876075

ABSTRACT

The heparan sulfate proteoglycan 2 (HSPG2)/perlecan gene is ancient and conserved in all triploblastic species. Its presence maintains critical cell boundaries in tissue and its large (up to ~900 kDa) modular structure has prompted speculation about the evolutionary origin of the gene. The gene's conservation amongst basal metazoans is unclear. After the recent sequencing of their genomes, the cnidarian Nematostella vectensis and the placozoan Trichoplax adhaerens have become favorite models for studying tissue regeneration and the evolution of multicellularity. More ancient basal metazoan phyla include the poriferan and ctenophore, whose evolutionary relationship has been clarified recently. Our in silico and PCR-based methods indicate that the HSPG2 gene is conserved in both the placozoan and cnidarian genomes, but not in those of the ctenophores and only partly in poriferan genomes. HSPG2 also is absent from published ctenophore and Capsaspora owczarzaki genomes. The gene in T. adhaerens is encoded as two separate but genetically juxtaposed genes that house all of the constituent pieces of the mammalian HSPG2 gene in tandem. These genetic constituents are found in isolated genes of various poriferan species, indicating a possible intronic recombinatory mechanism for assembly of the HSPG2 gene. Perlecan's expression during wound healing and boundary formation is conserved, as expression of the gene was activated during tissue regeneration and reformation of the basement membrane of N. vectensis. These data indicate that the complex HSPG2 gene evolved concurrently in a common ancestor of placozoans, cnidarians and bilaterians, likely along with the development of differentiated cell types separated by acellular matrices, and is activated to reestablish these tissue borders during wound healing.


Subject(s)
Cnidaria/genetics , Ctenophora/genetics , Heparan Sulfate Proteoglycans/genetics , Placozoa/genetics , Porifera/genetics , Regeneration/genetics , Amino Acid Sequence , Animals , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Cnidaria/classification , Cnidaria/metabolism , Cnidaria/ultrastructure , Ctenophora/classification , Ctenophora/metabolism , Ctenophora/ultrastructure , Evolution, Molecular , Gene Expression , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/metabolism , Humans , Models, Genetic , Molecular Sequence Data , Phylogeny , Placozoa/classification , Placozoa/metabolism , Placozoa/ultrastructure , Polymerase Chain Reaction , Porifera/classification , Porifera/metabolism , Porifera/ultrastructure , Sequence Alignment , Sequence Homology, Amino Acid
18.
J Cell Biochem ; 115(7): 1322-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24700612

ABSTRACT

Perlecan/HSPG2, a heparan sulfate proteoglycan typically found at tissue borders including those separating epithelia and connective tissue, increases near sites of invasion of primary prostatic tumors as previously shown for other proteins involved in desmoplastic tissue reaction. Studies of prostate cancer cells and stromal cells from both prostate and bone, the major site for prostate cancer metastasis, showed that cancer cells and a subset of stromal cells increased production of perlecan in response to cytokines present in the tumor microenvironment. In silico analysis of the HSPG2 promoter revealed two conserved NFκB binding sites, in addition to the previously reported SMAD3 binding sites. By systematically transfecting cells with a variety of reporter constructs including sequences up to 2.6 kb from the start site of transcription, we identified an active cis element in the distal region of the HSPG2 promoter, and showed that it functions in regulating transcription of HSPG2. Treatment with TNF-α and/or TGFß1 identified TNF-α as a major cytokine regulator of perlecan production. TNF-α treatment also triggered p65 nuclear translocation and binding to the HSPG2 regulatory region in stromal cells and cancer cells. In addition to stromal induction of perlecan production in the prostate, we identified a matrix-secreting bone marrow stromal cell type that may represent the source for increases in perlecan in the metastatic bone marrow environment. These studies implicate perlecan in cytokine-mediated, innate tissue responses to cancer cell invasion, a process we suggest reflects a modified wound healing tissue response co-opted by prostate cancer cells.


Subject(s)
Heparan Sulfate Proteoglycans/biosynthesis , Prostatic Neoplasms/genetics , Stromal Cells/cytology , Transcription Factor RelA/metabolism , Transcriptional Activation , Active Transport, Cell Nucleus , Binding Sites , Cell Line, Tumor , DNA-Binding Proteins , Heparan Sulfate Proteoglycans/genetics , Humans , Male , Promoter Regions, Genetic , Prostate/metabolism , Transforming Growth Factor beta1/pharmacology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
19.
Matrix Biol ; 34: 64-79, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24001398

ABSTRACT

The extracellular matrix proteoglycan (ECM) perlecan, also known as heparan sulfate proteoglycan 2 or HSPG2, is one of the largest (>200 nm) and oldest (>550 M years) extracellular matrix molecules. In vertebrates, perlecan's five-domain structure contains numerous independently folding modules with sequence similarities to other ECM proteins, all connected like cars into one long, diverse complex train following a unique N-terminal domain I decorated with three long glycosaminoglycan chains, and an additional glycosaminoglycan attachment site in the C-terminal domain V. In lower invertebrates, perlecan is not typically a proteoglycan, possessing the majority of the core protein modules, but lacking domain I where the attachment sites for glycosaminoglycan chains are located. This suggests that uniting the heparan sulfate binding growth factor functions of domain I and the core protein functions of the rest of the molecule in domains II-V occurred later in evolution for a new functional purpose. In this review, we surveyed several decades of pertinent literature to ask a fundamental question: Why did nature design this protein uniquely as an extraordinarily long multifunctional proteoglycan with a single promoter regulating expression, rather than separating these functions into individual proteins that could be independently regulated? We arrived at the conclusion that the concentration of perlecan at functional borders separating tissues and tissue layers is an ancient key function of the core protein. The addition of the heparan sulfate chains in domain I likely occurred as an additional means of binding the core protein to other ECM proteins in territorial matrices and basement membranes, and as a means to reserve growth factors in an on-site depot to assist with rapid repair of those borders when compromised, such as would occur during wounding. We propose a function for perlecan that extends its role from that of an extracellular scaffold, as we previously suggested, to that of a critical agent for establishing and patrolling tissue borders in complex tissues in metazoans. We also propose that understanding these unique functions of the individual portions of the perlecan molecule can provide new insights and tools for engineering of complex multi-layered tissues including providing the necessary cues for establishing neotissue borders.


Subject(s)
Basement Membrane/metabolism , Extracellular Matrix/metabolism , Heparan Sulfate Proteoglycans/metabolism , Protein Structure, Tertiary/genetics , Amino Acid Sequence/genetics , Animals , Basement Membrane/growth & development , Basement Membrane/ultrastructure , Extracellular Matrix/genetics , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Developmental , Heparan Sulfate Proteoglycans/genetics , Humans , Tissue Distribution/genetics
20.
Prostate ; 74(2): 149-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24122957

ABSTRACT

BACKGROUND: Bone marrow stromal cell (BMSC) paracrine factor(s) can induce apoptosis in bone metastatic prostate cancer (PCa) cell lines. However, the PCa cells that escape BMSC-induced apoptosis can upregulate cytoprotective autophagy. METHODS: C4-2, C4-2B, MDA PCa 2a, MDA PCa 2b, VCaP, PC3, or DU145 PCa cell lines were grown in BMSC conditioned medium and analyzed for mRNA and/or protein accumulation of p62 (also known as sequestome-1/SQSTM1), Microtubule-associated protein 1 light chain 3B (LC3B), or lysosomal-associated membrane protein 1 (LAMP1) using quantitative polymerase chain reaction (QPCR), Western blot, or immunofluorescence. Small interfering RNA (siRNA) was used to determine if p62 is necessary PCa cell survival. RESULTS: BMSC paracrine signaling upregulated p62 mRNA and protein in a subset of the PCa cell lines. The PCa cell lines that were insensitive to BMSC-induced apoptosis and autophagy induction had elevated basal p62 mRNA and protein. In the BMSC-insensitive PCa cell lines, siRNA knockdown of p62 was cytotoxic and immunostaining showed peri-nuclear clustering of autolysosomes. However, in the BMSC-sensitive PCa cell lines, p62 siRNA knockdown was not appreciably cytotoxic and did not affect autolysosome subcellular localization. CONCLUSIONS: A pattern emerges wherein the BMSC-sensitive PCa cell lines are known to be osteoblastic and express the androgen receptor, while the BMSC-insensitive PCa cell lines are characteristically osteolytic and do not express the androgen receptor. Furthermore, BMSC-insensitive PCa may have evolved a dependency on p62 for cell survival that could be exploited to target and kill these apoptosis-resistant PCa cells in the bone.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis/physiology , Bone Neoplasms/secondary , Prostatic Neoplasms/pathology , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/physiology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Neoplasm Invasiveness/pathology , Sequestosome-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...